Open Access

Associations of tumor suppressor SPARCL1 with cancer progression and prognosis (Review)

  • Authors:
    • Ting Li
    • Xia Liu
    • Antai Yang
    • Wenjie Fu
    • Fuqiang Yin
    • Xiaoyun Zeng
  • View Affiliations

  • Published online on: July 8, 2017     https://doi.org/10.3892/ol.2017.6546
  • Pages: 2603-2610
  • Copyright: © Li et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

SPARC‑like protein 1 (SPARCL1), a member of the family of secreted proteins which is acidic and rich in cysteine, is a potential tumor suppressor gene in most types of tumor. A systemic review and bioinformatics analysis was carried out to determine the associations between SPARCL1 and tumor progression and clinical factors. Downregulation of SPARCL1, thought to be regulated by epigenetic modifications including DNA methylation, serves important functions in tumor progression and development, with its regulatory functions on cell viability, migration, invasion, cell adhesion and drug resistance. Downregulation of SPARCL1 was markedly associated with a poor overall survival rate of patients with one of ≥7 solid tumors and predicted increased mortality in patients with one of ≥4 distinct tumor types. The present review indicated that SPARCL1 may be a therapeutic target for cancer treatment and a biomarker to determine prognosis.

SPARC-like protein 1 (SPARCL1) is a potential tumor suppressor gene

SPARCL1, a member of the family of secreted proteins that are acidic and rich in cysteine in the cellular matrix. Originally termed SC1, SPARCL1 was first cloned from the rat central nervous system and encodes an extracellular matrix glycoprotein, similar to osteonectin/basement membrane protein 40/secreted protein acidic and rich in cysteine (SPARC) (1). Subsequently, Schraml et al (2) and Girard et al (3) cloned the aforementioned gene from endothelial cells in non-small cell lung cancer and high endothelial venules in human tonsil lymphatic tissues, and termed it MAST9 and hevin, respectively. The mRNA of the gene is 3 kb in length and the theoretical molecular mass of the encoded protein, SPARCL1, is ~75 kDa. However, the protein expressed in vitro reveals molecular masses of ~75 and 150 kDa, suggesting that SPARCL1 protein may form a homodimer in vitro (4).

SPARC is a tumor suppressor gene in cancer, and it has been demonstrated to be involved in the regulation of tumor progression and drug resistance (5,6). SPARCL1 exhibits 62% identity with SPARC and the two proteins share three conservative structural domains (3), indicating functional similarity. SPARCL1 is localized on human chromosome 4, which contains a number of additional known tumor suppressor genes. Thus, SPARCL1 is considered to be a potential tumor suppressor gene and participates in tumor occurrence and development, by regulating tumor cell viability and differentiation (4). SPARCL1 may, additionally, be a potential oncogene and participates in tumor occurrence and development, by regulating tumor cell viability and affecting the production of tumor blood vessels (7).

In the present review, the mRNA expression of SPARCL1 in tumors was analyzed using Oncomine (www.oncomine.org/resource/login.html) (8). As presented in Fig. 1, of the ~20 different types of solid tumors included in the Oncomine database, SPARCL1 was downregulated >2-fold in the majority of tumors analyzed, with the exception of liver cancer, lymphoma and sarcoma, where SPARCL1 was upregulated. The downregulation of SPARCL1 was marked in bladder, breast, cervical, rectal, lung and ovarian cancer. Thus, it may be inferred that SPARCL1 is a tumor suppressor gene in cancer.

DNA methylation may be an important mechanism that contributes to the downregulation of SPARCL1

As presented in Table I, SPARCL1 is upregulated in liver cancer (7); however, SPARCL1 is markedly downregulated in prostate (9), lung (10), ovarian (11) and a number of other types of cancer. These results are consistent with Fig. 1. Downregulation of SPARCL1 in tumors may result from the epigenetic mechanisms, including DNA methylation, because SPARCL1 is not a classical tumor suppressor gene exhibiting a deletion or mutation. Isler et al (10) used microsatellite analysis, quantitative polymerase chain reaction and sequence analysis of all exons, including the intron-exon junctions and a portion of the putative promoter region, but did not identify a mutation or deletion that may be responsible for the downregulation of SPARCL1. This was indicative of other regulatory mechanisms resulting in the differential expression of SPARCL1 in tumors, including epigenetic modification. A previous study revealed that DNA methylation is the reason for the downregulation of SPARCL1 in pancreatic, ovarian and lung cancers (Table I), and demethylation of the gene partially reversed the abnormal expression in pancreatic cancer (12).

Table I.

Summary on the SPARCL1 correlated with tumor progression and development.

Table I.

Summary on the SPARCL1 correlated with tumor progression and development.

Tumor typeSPARCL1 expressionBiological functionMolecular mechanismMechanism of gene expression
Prostate cancerDownregulation (9)Inhibition of cell migration and invasion (14)Affects migration by regulating RhoC (19); inhibition of the assembly of focal adhesions (20)
Colorectal cancerDownregulation (13,21)Inhibition of cell proliferation, growth and invasion (13)Affects tumor cell differentiation through EMT (13)
Hilar cholangiocarcinomaDownregulation (15)Inhibition of cell migration (15)Inhibition of the expression of MMP-9, MMP-2, vimentin and fibronectin (15)
Gastric cancerDownregulation (22,23)Inactivation of its tumor suppressor functions (22)Loss of heterozygosity (23)
Pancreatic cancerDownregulation (12)Inhibition of tumor cell proliferation and invasion (12)Anti-invasive effects (12)DNA methylation (12)
Ovarian cancerDownregulation (11)Associated with multiple drug resistance (18)Interaction with drug resistance-related proteins including PTEN (18)DNA methylation (24)
Lung cancerDownregulation (10)Decrease in luciferase activity (10)DNA methylation (25)
GliomaDownregulation (16)Associated with cell cycle (16)Anti-adhesion (16)
Liver cancerUpregulation (7)
Uterine leiomyomaUpregulation (26)

[i] SPARCL1, SPARC-like protein 1; MMP, matrix metalloproteinase; PTEN, phosphatase and tensin homolog.

SPARCL1 contributes to tumor development and progression

There have been a limited number of studies on SPARCL1, but the gene has been identified to be markedly associated with tumor development and progression. SPARCL1 contributions to tumor cell viability (13), migration and invasion (1216) and exhibits an anti-adhesive effect (12,16). In addition, SPARCL1 may be involved in the regulation of drug resistance in cancer. It has been identified that SPARCL1 is a recombinant gene in the extracellular matrix of osteosarcoma in children and is involved in the mechanism of multiple drug resistance (17). A previous study used comprehensive bioinformatics analysis to identify that the SPARCL1 gene was involved in the regulation of drug resistance in ovarian cancer (18).

The association between SPARCL1 and tumor progression was investigated using Coremine Medical (http://www.coremine.com/medical). As presented in Fig. 2, using SPARCL1 and cancer as key words, SPARCL1 was identified to be associated with diagnosis, prognosis, recurrence, invasiveness, metastasis and drug resistance of cancer (Fig. 2A). In addition, the associations identified in the present review, between SPARCL1 and invasiveness, metastasis and drug resistance of cancer, were consistent with previous studies (Table I)(916,1826). Furthermore, analysis indicated that SPARCL1 may participate in cancer development and progression, in 9 biological processes (P<0.001) including cell viability, cell cycle, migration and adhesion (Fig. 2B), which is consistent with previous studies (Table I). In addition, SPARCL1 and cancer were annotated with DNA methylation, supporting the hypothesis that DNA methylation may be an important mechanism which contributes to the downregulation of SPARCL1.

Downregulation of SPARCL1 is associated with poor prognosis in cancer

Previous studies indicate that downregulation of SPARCL1 is markedly associated with poor prognosis and therefore the gene may be a prognostic marker in cancers. In prostate cancer, the downregulation of SPARCL1 has been markedly associated with biochemical recurrence, metastatic disease and poor overall survival (OS) time (19). Patients with stage II/III colorectal cancer who possessed increased p53 and decreased SPARCL1 expression levels exhibited ~50% decreased 3-year survival compared with controls (27). Furthermore, in gastric cancer, silenced expression of SPARCL1 predicted a poorer prognosis (23).

On the basis of The Cancer Genome Atlas (TCGA) (24) cohort data, the associations between SPARCL1 and cancer prognosis were analyzed. The expression value of SPARCL1 and the corresponding clinical data of each type of cancer in the TCGA cohort was retrieved from the cBioPortal database (cbioportal.org) (28). Expression values of SPARCL1 were divided into high and low expression using the median as the threshold value in a Kaplan-Meier estimator analysis, in accordance with a previous study (29). As presented in Table II and Fig. 3, downregulation of SPARCL1 was markedly associated with poor OS time in liver cancer (242 samples) and lung cancer (324 samples), and markedly associated with poorer disease-free survival and OS time in glioma (311 samples).

Table II.

Association between SPARCL1 expression and prognosis in glioma, lung cancer and liver cancer.

Table II.

Association between SPARCL1 expression and prognosis in glioma, lung cancer and liver cancer.

DFS (median)OS (median)


95% confidence interval 95% confidence interval


Disease typeGroupEstimateStandard errorLower boundaryUpper boundaryEstimateStandard errorLower boundaryUpper boundary
GliomaL37.900   3.98330.09345.70762.90012.97137.476   88.324
H62.00010.17342.06181.93998.20019.62759.731136.669
Total43.500   4.52434.63352.36787.40010.23267.346107.454
Lung cancerL 42.500   3.92034.818   50.182
H 60.10012.42535.747   84.453
Total 49.200   3.70541.938   56.462
Liver cancerL 40.400   9.14222.483   58.317
H 80.70010.85359.428101.972
Total 55.600   8.08739.749   71.451

[i] SPARCL1 expression and survival data in The Cancer Genome Atlas cohort were used for the analysis. Expression values of a gene were divided into high and low expression using the median as a cut-off. H, high expression; L, low expression; DFS, disease-free survival; OS, overall survival.

The association between SPARCL1 and OS time in lung cancer was additionally validated using Kaplan-Meier estimator analysis, which selected thousands of samples of ovarian, lung, breast and gastric cancer from microarrays deposited in the TCGA cohort and Gene Expression Omnibus profiles (30). Using the median expression as the threshold value, it was identified that the downregulation of SPARCL1 in lung cancer (3,021 samples) predicted decreased OS time (Fig. 4A), which was consistent with the results based on the TCGA cohort data (Table II and Fig. 3). Furthermore, downregulation of SPARCL1 predicted improved OS time in gastric cancer (1,223 samples) (Fig. 4B), although this result was in contrast with a previous study (23). In addition, in breast cancer (2,627 samples), downregulation of SPARCL1 predicted poorer OS time (Fig. 4C).

SPARCL1 was additionally associated with clinical features of a number of types of tumor. Downregulation of SPARCL1 was associated with increased mortality of patients with glioma, liver and lung cancer (P<0.05), and patients with cervical cancer (P=0.076; Table III). Furthermore, the downregulation rate of SPARCL1 increased considerably for surviving patients with cervical cancer and downregulation of the gene in ovarian cancer was markedly associated with a lower histological grade (P<0.05; Table IV).

Table III.

Association between SPARCL1 expression and vital status of patients with different tumors, in accordance with The Cancer Genome Atlas cohort.

Table III.

Association between SPARCL1 expression and vital status of patients with different tumors, in accordance with The Cancer Genome Atlas cohort.

SPARCL1 expression

Patient's vital statusNo. of patientsLow (%)High (%)P-value
Glioma of lower grade528 0.001
  Deceased134 (25.4%)  84 (62.7)  50 (37.3)
  Alive394 (74.6%)180 (45.7)214 (54.3)
Lung adenocarcinoma516 0.001
  Deceased187 (36.2%)111 (59.4)  76 (40.6)
  Alive329 (63.8%)146 (44.4)183 (55.6)
Hepatocellular carcinoma372 0.024
  Deceased130 (34.9%)  75 (57.7)  55 (42.3)
  Alive242 (65.1%)110 (45.5)132 (54.5)
Cervical adenocarcinoma305 0.076
  Deceased73 (23.9%)  43 (58.9)  30 (41.1)
  Alive232 (76.1%)109 (47.0)123 (53.0)
Subcutaneous melanoma470 0.42
  Deceased222 (47.2%)100 (45.0)122 (55.0)
  Alive248 (52.8%)135 (54.4)113 (45.6)
Acute myeloid leukemia173 0.916
  Deceased114 (65.9%)  57 (50.0)  57 (50.0)
  Alive59 (34.1%)  29 (49.2)  30 (50.8)
Lymphoma27 1
  Deceased6 (22.2%)     3 (50.0)     3 (50.0)
  Alive21 (77.8%)  11 (52.4)  10 (47.6)
Prostate adenocarcinoma497 0.339
  Deceased10 (2.0%)     7 (70.0)     3 (30.0)
  Alive487 (98.0%)242 (49.7)245 (50.3)
Sarcoma261 0.666
  Deceased99 (37.9%)  51 (51.5)  48 (48.5)
  Alive162 (62.1%)  79 (48.8)  83 (51.2)
Esophageal carcinoma184 0.981
  Deceased77 (41.8%)  38 (49.4)  39 (50.6)
  Alive107 (58.2%)  53 (49.5)  54 (50.5)

[i] Expression values of SPARCL1 were divided into high and low expression using the median as the threshold value. Cervical adenocarcinoma is defined as a cervical squamous cell carcinoma and endocervical adenocarcinoma. Lymphoma is defined as a lymphoid neoplasm diffuse large B-cell lymphoma. SPARCL1, SPARC-like protein 1.

Table IV.

Association of SPARCL1 expression with neoplasm status and neoplasm histological grade in several cancers, in accordance with The Cancer Genome Atlas cohort.

Table IV.

Association of SPARCL1 expression with neoplasm status and neoplasm histological grade in several cancers, in accordance with The Cancer Genome Atlas cohort.

SPARCL1 expression

VariablesNo. of patientsLow (%)High (%)P-value
Glioma of lower grade442 0.036
  With tumor220 (49.8%)100 (45.5)120 (54.5)
  Tumor-free222 (50.2%)123 (55.4)  99 (44.6)
Cervical adenocarcinoma263 0.013
  With tumor76 (28.9%)  47 (61.8)  29 (38.2)
  Tumor-free187 (71.1%)  84 (44.9)103 (50.2)
Ovarian serous cystadenocarcinoma476 0.004
  Histological grade 256 (11.8%)  18 (32.1)  38 (67.9)
  Histological grade 3420 (88.2%)220 (52.4)200 (47.6)

[i] Expression values of SPARCL1 were divided into high and low expression using the median as the threshold value. Cervical adenocarcinoma is defined as a cervical squamous cell carcinoma and endocervical adenocarcinoma.

Conclusions

Previous studies on the association between SPARCL1 and tumor progression are relatively limited. One previous study suggested that SPARCL1 is an oncogene (7), but a number of contradictory studies have identified SPARCL1 as a potential tumor suppressor gene (4,12,13,15). A bioinformatic analysis, on the basis of the data retrieved from Oncomine and the TCGA cohort, was conducted to identify the associations between SPARCL1 and tumor progression. Oncomine included information concerning SPARCL1 expression in almost 20 solid tumors (Fig. 1) and this identified that downregulation of SPARCL1 is prevalent in the majority of tumors, suggesting that SPARCL1 is a tumor suppressor gene.

The present review revealed that the downregulation of the SPARCL1 was markedly associated with poor OS time of ≥7 solid tumors, which included prostate (19), colorectal (27), gastric (23), liver, lung, glioma and breast cancer (Table II; Figs. 3 and 4). In addition, decreased expression of SPARCL1 typically predicted increased mortality in glioma, lung, liver and cervical cancer (Table III). Therefore, SPARCL1 may be a universal prognostic marker of tumors in the clinic.

Previous studies have indicated that SPARCL1 is a tumor suppressor gene and is involved in tumor cell viability (13), migration and invasion (1216) and cell adhesion (12,16), and is associated with drug resistance of tumors (17,18). Consistent with these studies, bioinformatics analysis in the present review revealed that SPARCL1 was associated with prognosis, invasiveness, metastasis, recurrence and drug resistance of cancer (Fig. 2A). It is hypothesized that SPARCL1 exhibits these aforementioned actions by interactions with a number of biological processes/signaling pathways including cell adhesion, cell viability, cell cycle and cell migration (Fig. 2B). These results indicate that SPARCL1 serves important functions in tumor progression.

The present review has elucidated the association between SAPRCL1 and cancer. SPARCL1 may be an important tumor suppressor gene in tumor progression and development, and it may be a therapeutic target for cancer treatment and a potential biomarker for prognosis.

Acknowledgements

The present review was supported by the National Natural Science Foundation of China (grant nos. 81302283, 81560424, 81660606 and 81460397), the China Postdoctoral Science Foundation (grant nos. 2014M552535XB and 2014M552291) and the Natural Science Foundation of Guangxi (grant nos. 2014GXNSFCA118010, 2015GXNSFBA139115, 2015GXNSFAA139151 and 2014GXNSFBA118155).

References

1 

Johnston IG, Paladino T, Gurd JW and Brown IR: Molecular cloning of SC1: A putative brain extracellular matrix glycoprotein showing partial similarity to osteonectin/BM40/SPARC. Neuron. 4:165–176. 1990. View Article : Google Scholar : PubMed/NCBI

2 

Schraml P, Shipman R, Stulz P and Ludwig CU: cDNA subtraction library construction using a magnet-assisted subtraction technique (MAST). Trends Genet. 9:70–71. 1993. View Article : Google Scholar : PubMed/NCBI

3 

Girard JP and Springer TA: Cloning from purified high endothelial venule cells of hevin, a close relative of the antiadhesive extracellular matrix protein SPARC. Immunity. 2:113–123. 1995. View Article : Google Scholar : PubMed/NCBI

4 

Claeskens A, Ongenae N, Neefs JM, Cheyns P, Kaijen P, Cools M and Kutoh E: Hevin is down-regulated in many cancers and is a negative regulator of cell growth and proliferation. Br J Cancer. 82:1123–1130. 2000. View Article : Google Scholar : PubMed/NCBI

5 

Socha MJ, Said N, Dai Y, Kwong J, Ramalingam P, Trieu V, Desai N, Mok SC and Motamed K: Aberrant promoter methylation of SPARC in ovarian cancer. Neoplasia. 11:126–135. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Tai IT, Dai M, Owen DA and Chen LB: Genome-wide expression analysis of therapy-resistant tumors reveals SPARC as a novel target for cancer therapy. J Clin Invest. 115:1492–1502. 2005. View Article : Google Scholar : PubMed/NCBI

7 

Lau CP, Poon RT, Cheung ST, Yu WC and Fan ST: SPARC and Hevin expression correlate with tumour angiogenesis in hepatocellular carcinoma. J Pathol. 210:459–468. 2006. View Article : Google Scholar : PubMed/NCBI

8 

Rhodes DR, Yu J, Shanker K, Deshpande N, Varambally R, Ghosh D, Barrette T and Pandey A: ONCOMINE: A cancer microarray database and integrated data-mining platform. Neoplasia. 6:1–6. 2004. View Article : Google Scholar : PubMed/NCBI

9 

Schlomm T, Luebke AM, Sültmann H, Hellwinkel OJ, Sauer U, Poustka A, David KA, Chun FK, Haese A, Graefen M, et al: Extraction and processing of high quality RNA from impalpable and macroscopically invisible prostate cancer for microarray gene expression analysis. Int J Oncol. 27:713–720. 2005.PubMed/NCBI

10 

Isler SG, Ludwig CU, Chiquet-Ehrismann R and Schenk S: Evidence for transcriptional repression of SPARC-like 1, a gene downregulated in human lung tumors. Int J Oncol. 25:1073–1079. 2004.PubMed/NCBI

11 

Biade S, Marinucci M, Schick J, Roberts D, Workman G, Sage EH, O'Dwyer PJ, Livolsi VA and Johnson SW: Gene expression profiling of human ovarian tumours. Br J Cancer. 95:1092–1100. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Esposito I, Kayed H, Keleg S, Giese T, Sage EH, Schirmacher P, Friess H and Kleeff J: Tumor-suppressor function of SPARC-like protein 1/hevin in pancreatic cancer. Neoplasia. 9:8–17. 2007. View Article : Google Scholar : PubMed/NCBI

13 

Hu H, Zhang H, Ge W, Liu X, Loera S, Chu P, Chen H, Peng J, Zhou L, Yu S, et al: Secreted protein acidic and rich in cysteines-like 1 suppresses aggressiveness and predicts better survival in colorectal cancers. Clin Cancer Res. 18:5438–5448. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Xiang Y, Qiu Q, Jiang M, Jin R, Lehmann BD, Strand DW, Jovanovic B, DeGraff DJ, Zheng Y, Yousif DA, et al: SPARCL1 suppresses metastasis in prostate cancer. Mol Oncol. 7:1019–1030. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Yu Y, Chen Y, Ma J, Yu X, Yu G and Li Z: SPARCL1 is a novel predictor of tumor recurrence and survival in hilar cholangiocarcinoma. Tumour Biol. 37:4159–4167. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Turtoi A, Musmeci D, Naccarato AG, Scatena C, Ortenzi V, Kiss R, Murtas D, Patsos G, Mazzucchelli G, De Pauw E, et al: Sparc-like protein 1 is a new marker of human glioma progression. J Proteome Res. 11:5011–5021. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Mintz MB, Sowers R, Brown KM, Hilmer SC, Mazza B, Huvos AG, Meyers PA, Lafleur B, McDonough WS, Henry MM, et al: An expression signature classifies chemotherapy-resistant pediatric osteosarcoma. Cancer Res. 65:1748–1754. 2005. View Article : Google Scholar : PubMed/NCBI

18 

Yin F, Liu X, Li D, Wang Q, Zhang W and Li L: Bioinformatic analysis of chemokine (C-C motif) ligand 21 and SPARC-like protein 1 revealing their associations with drug resistance in ovarian cancer. Int J Oncol. 42:1305–1316. 2013.PubMed/NCBI

19 

Hurley PJ, Marchionni L, Simons BW, Ross AE, Peskoe SB, Miller RM, Erho N, Vergara IA, Ghadessi M, Huang Z, et al: Secreted protein, acidic and rich in cysteine-like 1 (SPARCL1) is down regulated in aggressive prostate cancers and is prognostic for poor clinical outcome. Proc Natl Acad Sci USA. 109:14977–14982. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Hurley PJ, Hughes RM, Simons BW, Huang J, Miller RM, Shinder B, Haffner MC, Esopi D, Kimura Y, Jabbari J, et al: Androgen-Regulated SPARCL1 in the Tumor Microenvironment Inhibits Metastatic Progression. Cancer Res. 75:4322–4334. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Zhang H, Widegren E, Wang DW and Sun XF: SPARCL1: A potential molecule associated with tumor diagnosis, progression and prognosis of colorectal cancer. Tumour Biol. 32:1225–1231. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Jakharia A, Borkakoty B and Singh S: Expression of SPARC like protein 1 (SPARCL1), extracellular matrix-associated protein is down regulated in gastric adenocarcinoma. J Gastrointest Oncol. 7:278–283. 2016.PubMed/NCBI

23 

Li P, Qian J, Yu G, Chen Y, Liu K, Li J and Wang J: Down-regulated SPARCL1 is associated with clinical significance in human gastric cancer. J Surg Oncol. 105:31–37. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Cancer Genome Atlas Research Network: Integrated genomic analyses of ovarian carcinoma. Nature. 474:609–615. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Sato T, Arai E, Kohno T, Takahashi Y, Miyata S, Tsuta K, Watanabe S, Soejima K, Betsuyaku T and Kanai Y: Epigenetic clustering of lung adenocarcinomas based on DNA methylation profiles in adjacent lung tissue: Its correlation with smoking history and chronic obstructive pulmonary disease. Int J Cancer. 135:319–334. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Mencalha AL, Levinsphul A, Deterling LC, Pizzatti L and Abdelhay E: SPARC-like1 mRNA is overexpressed in human uterine leiomyoma. Mol Med Rep. 1:571–574. 2008.PubMed/NCBI

27 

Yu SJ, Yu JK, Ge WT, Hu HG, Yuan Y and Zheng S: SPARCL1, Shp2, MSH2, E-cadherin, p53, ADCY-2 and MAPK are prognosis-related in colorectal cancer. World J Gastroenterol. 17:2028–2036. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, Sun Y, Jacobsen A, Sinha R, Larsson E, et al: Integrative Analysis of Complex Cancer Genomics and Clinical Profiles Using the cBioPortal. Sci Signal. 6:pl12013. View Article : Google Scholar : PubMed/NCBI

29 

Hedditch EL, Gao B, Russell AJ, Lu Y, Emmanuel C, Beesley J, Johnatty SE, Chen X, Harnett P, George J, et al: ABCA transporter gene expression and poor outcome in epithelial ovarian cancer. J Natl Cancer Inst. 106:pii: dju149. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Gyorffy B, Lánczky A and Szállási Z: Implementing an online tool for genome-wide validation of survival-associated biomarkers in ovarian-cancer using microarray data from 1287 patients. Endocr Relat Cancer. 19:197–208. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2017
Volume 14 Issue 3

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li T, Liu X, Yang A, Fu W, Yin F and Zeng X: Associations of tumor suppressor SPARCL1 with cancer progression and prognosis (Review). Oncol Lett 14: 2603-2610, 2017
APA
Li, T., Liu, X., Yang, A., Fu, W., Yin, F., & Zeng, X. (2017). Associations of tumor suppressor SPARCL1 with cancer progression and prognosis (Review). Oncology Letters, 14, 2603-2610. https://doi.org/10.3892/ol.2017.6546
MLA
Li, T., Liu, X., Yang, A., Fu, W., Yin, F., Zeng, X."Associations of tumor suppressor SPARCL1 with cancer progression and prognosis (Review)". Oncology Letters 14.3 (2017): 2603-2610.
Chicago
Li, T., Liu, X., Yang, A., Fu, W., Yin, F., Zeng, X."Associations of tumor suppressor SPARCL1 with cancer progression and prognosis (Review)". Oncology Letters 14, no. 3 (2017): 2603-2610. https://doi.org/10.3892/ol.2017.6546